Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Mitochondria as multifaceted regulators of cell death

Abstract

Through their many and varied metabolic functions, mitochondria power life. Paradoxically, mitochondria also have a central role in apoptotic cell death. Upon induction of mitochondrial apoptosis, mitochondrial outer membrane permeabilization (MOMP) usually commits a cell to die. Apoptotic signalling downstream of MOMP involves cytochrome c release from mitochondria and subsequent caspase activation. As such, targeting MOMP in order to manipulate cell death holds tremendous therapeutic potential across different diseases, including neurodegenerative diseases, autoimmune disorders and cancer. In this Review, we discuss new insights into how mitochondria regulate apoptotic cell death. Surprisingly, recent data demonstrate that besides eliciting caspase activation, MOMP engages various pro-inflammatory signalling functions. As we highlight, together with new findings demonstrating cell survival following MOMP, this pro-inflammatory role suggests that mitochondria-derived signalling downstream of pro-apoptotic cues may also have non-lethal functions. Finally, we discuss the importance and roles of mitochondria in other forms of regulated cell death, including necroptosis, ferroptosis and pyroptosis. Collectively, these new findings offer exciting, unexplored opportunities to target mitochondrial regulation of cell death for clinical benefit.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Apoptotic signalling pathways.
Fig. 2: BAX/BAK-mediated mitochondrial outer membrane permeabilization.
Fig. 3: Differential levels of mitochondrial outer membrane permeabilization permit cell survival and unmask signalling functions.
Fig. 4: Pro-inflammatory effects of mitochondrial outer membrane permeabilization.
Fig. 5: Inhibition of mitochondrial outer membrane permeabilization-induced inflammation.
Fig. 6: Mitochondria and non-apoptotic cell death.
Fig. 7: Strategies to target mitochondrial apoptosis in disease.

Similar content being viewed by others

References

  1. Mehta, M. M., Weinberg, S. E. & Chandel, N. S. Mitochondrial control of immunity: beyond ATP. Nat. Rev. Immunol. 17, 608–620 (2017).

    CAS  PubMed  Google Scholar 

  2. Filippi, M. D. & Ghaffari, S. Mitochondria in the maintenance of hematopoietic stem cells: new perspectives and opportunities. Blood 133, 1943–1952 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Merino, D. et al. BH3-mimetic drugs: blazing the trail for new cancer medicines. Cancer Cell 34, 879–891 (2018).

    CAS  PubMed  Google Scholar 

  4. Roberts, A. W. et al. Targeting BCL2 with venetoclax in relapsed chronic lymphocytic leukemia. N. Engl. J. Med. 374, 311–322 (2016).

    CAS  PubMed  Google Scholar 

  5. Tuzlak, S., Kaufmann, T. & Villunger, A. Interrogating the relevance of mitochondrial apoptosis for vertebrate development and postnatal tissue homeostasis. Genes Dev. 30, 2133–2151 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Julien, O. & Wells, J. A. Caspases and their substrates. Cell Death Differ. 24, 1380–1389 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Boatright, K. M. et al. A unified model for apical caspase activation. Mol. Cell 11, 529–541 (2003).

    CAS  PubMed  Google Scholar 

  8. Dorstyn, L., Akey, C. W. & Kumar, S. New insights into apoptosome structure and function. Cell Death Differ. 25, 1194–1208 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. McCarthy, N. J., Whyte, M. K., Gilbert, C. S. & Evan, G. I. Inhibition of Ced-3/ICE-related proteases does not prevent cell death induced by oncogenes, DNA damage, or the Bcl-2 homologue Bak. J. Cell Biol. 136, 215–227 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Xiang, J., Chao, D. T. & Korsmeyer, S. J. BAX-induced cell death may not require interleukin 1β-converting enzyme-like proteases. Proc. Natl Acad. Sci. USA 93, 14559–14563 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Amarante-Mendes, G. P. et al. Anti-apoptotic oncogenes prevent caspase-dependent and independent commitment for cell death. Cell Death Differ. 5, 298–306 (1998).

    CAS  PubMed  Google Scholar 

  12. Cecconi, F., Alvarez-Bolado, G., Meyer, B. I., Roth, K. A. & Gruss, P. Apaf1 (CED-4 homolog) regulates programmed cell death in mammalian development. Cell 94, 727–737 (1998).

    CAS  PubMed  Google Scholar 

  13. Yoshida, H. et al. Apaf1 is required for mitochondrial pathways of apoptosis and brain development. Cell 94, 739–750 (1998).

    CAS  PubMed  Google Scholar 

  14. Kuida, K. et al. Reduced apoptosis and cytochrome c-mediated caspase activation in mice lacking caspase 9. Cell 94, 325–337 (1998).

    CAS  PubMed  Google Scholar 

  15. Ke, F. F. S. et al. Embryogenesis and adult life in the absence of intrinsic apoptosis effectors BAX, BAK, and BOK. Cell 173, 1217–1230 e1217 (2018). This study affirms an important role for mitochondrial apoptosis in embryonic development but, surprisingly, shows that some apoptosis-deficient mice can survive to adulthood.

    CAS  PubMed  Google Scholar 

  16. Lindsten, T. et al. The combined functions of proapoptotic Bcl-2 family members bak and bax are essential for normal development of multiple tissues. Mol. Cell 6, 1389–1399 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Lakhani, S. A. et al. Caspases 3 and 7: key mediators of mitochondrial events of apoptosis. Science 311, 847–851 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Goldstein, J. C., Waterhouse, N. J., Juin, P., Evan, G. I. & Green, D. R. The coordinate release of cytochrome c during apoptosis is rapid, complete and kinetically invariant. Nat. Cell Biol. 2, 156–162 (2000).

    CAS  PubMed  Google Scholar 

  19. Lartigue, L. et al. Caspase-independent mitochondrial cell death results from loss of respiration, not cytotoxic protein release. Mol. Biol. Cell 20, 4871–4884 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Wei, M. C. et al. Proapoptotic BAX and BAK: a requisite gateway to mitochondrial dysfunction and death. Science 292, 727–730 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Sarosiek, K. A. et al. BID preferentially activates BAK while BIM preferentially activates BAX, affecting chemotherapy response. Mol. Cell 51, 751–765 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Lopez, J. et al. Mito-priming as a method to engineer Bcl-2 addiction. Nat. Commun. 7, 10538 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Lauterwasser, J. et al. The porin VDAC2 is the mitochondrial platform for Bax retrotranslocation. Sci. Rep. 6, 32994 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Naghdi, S., Varnai, P. & Hajnoczky, G. Motifs of VDAC2 required for mitochondrial Bak import and tBid-induced apoptosis. Proc. Natl Acad. Sci. USA 112, E5590–E5599 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Chin, H. S. et al. VDAC2 enables BAX to mediate apoptosis and limit tumor development. Nat. Commun. 9, 4976 (2018).

    PubMed  PubMed Central  Google Scholar 

  26. Edlich, F. et al. Bcl-xL retrotranslocates Bax from the mitochondria into the cytosol. Cell 145, 104–116 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Todt, F. et al. Differential retrotranslocation of mitochondrial Bax and Bak. EMBO J. 34, 67–80 (2015).

    CAS  PubMed  Google Scholar 

  28. Schellenberg, B. et al. Bax exists in a dynamic equilibrium between the cytosol and mitochondria to control apoptotic priming. Mol. Cell 49, 959–971 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Letai, A. et al. Distinct BH3 domains either sensitize or activate mitochondrial apoptosis, serving as prototype cancer therapeutics. Cancer Cell 2, 183–192 (2002).

    CAS  PubMed  Google Scholar 

  30. Czabotar, P. E. et al. Bax crystal structures reveal how BH3 domains activate Bax and nucleate its oligomerization to induce apoptosis. Cell 152, 519–531 (2013).

    CAS  PubMed  Google Scholar 

  31. Moldoveanu, T. et al. BID-induced structural changes in BAK promote apoptosis. Nat. Struct. Mol. Biol. 20, 589–597 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Leshchiner, E. S., Braun, C. R., Bird, G. H. & Walensky, L. D. Direct activation of full-length proapoptotic BAK. Proc. Natl Acad. Sci. USA 110, E986–E995 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Brouwer, J. M. et al. Conversion of Bim-BH3 from activator to inhibitor of Bak through structure-based design. Mol. Cell 68, 659–672 e659 (2017).

    CAS  PubMed  Google Scholar 

  34. Gavathiotis, E. et al. BAX activation is initiated at a novel interaction site. Nature 455, 1076–1081 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Gavathiotis, E., Reyna, D. E., Davis, M. L., Bird, G. H. & Walensky, L. D. BH3-triggered structural reorganization drives the activation of proapoptotic BAX. Mol. Cell 40, 481–492 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Reyna, D. E. et al. Direct activation of BAX by BTSA1 overcomes apoptosis resistance in acute myeloid leukemia. Cancer Cell 32, 490–505 e410 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Dengler, M. A. et al. BAX activation: mutations near its proposed non-canonical BH3 binding site reveal allosteric changes controlling mitochondrial association. Cell Rep. 27, 359–373 e356 (2019).

    CAS  PubMed  Google Scholar 

  38. Chen, H. C. et al. An interconnected hierarchical model of cell death regulation by the BCL-2 family. Nat. Cell Biol. 17, 1270–1281 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Dewson, G. et al. Bak activation for apoptosis involves oligomerization of dimers via their α6 helices. Mol. Cell 36, 696–703 (2009).

    CAS  PubMed  Google Scholar 

  40. Dewson, G. et al. To trigger apoptosis, Bak exposes its BH3 domain and homodimerizes via BH3:groove interactions. Mol. Cell 30, 369–380 (2008).

    CAS  PubMed  Google Scholar 

  41. Dewson, G. et al. Bax dimerizes via a symmetric BH3:groove interface during apoptosis. Cell Death Differ. 19, 661–670 (2012).

    CAS  PubMed  Google Scholar 

  42. Subburaj, Y. et al. Bax monomers form dimer units in the membrane that further self-assemble into multiple oligomeric species. Nat. Commun. 6, 8042 (2015).

    CAS  PubMed  Google Scholar 

  43. Bleicken, S. et al. Molecular details of Bax activation, oligomerization, and membrane insertion. J. Biol. Chem. 285, 6636–6647 (2010).

    CAS  PubMed  Google Scholar 

  44. Gillies, L. A. et al. Visual and functional demonstration of growing Bax-induced pores in mitochondrial outer membranes. Mol. Biol. Cell 26, 339–349 (2015).

    PubMed  PubMed Central  Google Scholar 

  45. Bleicken, S., Landeta, O., Landajuela, A., Basanez, G. & Garcia-Saez, A. J. Proapoptotic Bax and Bak proteins form stable protein-permeable pores of tunable size. J. Biol. Chem. 288, 33241–33252 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Salvador-Gallego, R. et al. Bax assembly into rings and arcs in apoptotic mitochondria is linked to membrane pores. EMBO J. 35, 389–401 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Grosse, L. et al. Bax assembles into large ring-like structures remodeling the mitochondrial outer membrane in apoptosis. EMBO J. 35, 402–413 (2016). Together with Salvador-Gallego et al. (2016), this work uses super-resolution microscopy to visualize, for the first time, BAX pores on the mitochondrial outer membrane.

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Llambi, F. et al. BOK is a non-canonical BCL-2 family effector of apoptosis regulated by ER-associated degradation. Cell 165, 421–433 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Einsele-Scholz, S. et al. Bok is a genuine multi-BH-domain protein that triggers apoptosis in the absence of Bax and Bak. J. Cell Sci. 129, 2213–2223 (2016). Together with Llambi et al. (2016), this study demonstrates that BOK can mediate MOMP and apoptosis in the absence of BAX and BAK.

    CAS  PubMed  Google Scholar 

  50. Fernandez-Marrero, Y. et al. The membrane activity of BOK involves formation of large, stable toroidal pores and is promoted by cBID. FEBS J. 284, 711–724 (2017).

    CAS  PubMed  Google Scholar 

  51. Zheng, J. H. et al. Intrinsic instability of BOK enables membrane permeabilization in apoptosis. Cell Rep. 23, 2083–2094 e2086 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Ke, F. et al. BCL-2 family member BOK is widely expressed but its loss has only minimal impact in mice. Cell Death Differ. 19, 915–925 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Wang, Y. et al. Chemotherapy drugs induce pyroptosis through caspase-3 cleavage of a gasdermin. Nature 547, 99–103 (2017).

    CAS  PubMed  Google Scholar 

  54. Rogers, C. et al. Cleavage of DFNA5 by caspase-3 during apoptosis mediates progression to secondary necrotic/pyroptotic cell death. Nat. Commun. 8, 14128 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Rogers, C. et al. Gasdermin pores permeabilize mitochondria to augment caspase-3 activation during apoptosis and inflammasome activation. Nat. Commun. 10, 1689 (2019).

    PubMed  PubMed Central  Google Scholar 

  56. Rehm, M., Dussmann, H. & Prehn, J. H. Real-time single cell analysis of Smac/DIABLO release during apoptosis. J. Cell Biol. 162, 1031–1043 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Lartigue, L. et al. An intracellular wave of cytochrome c propagates and precedes Bax redistribution during apoptosis. J. Cell Sci. 121, 3515–3523 (2008).

    CAS  PubMed  Google Scholar 

  58. Bhola, P. D., Mattheyses, A. L. & Simon, S. M. Spatial and temporal dynamics of mitochondrial membrane permeability waves during apoptosis. Biophys. J. 97, 2222–2231 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Rehm, M. et al. Dynamics of outer mitochondrial membrane permeabilization during apoptosis. Cell Death Differ. 16, 613–623 (2009).

    CAS  PubMed  Google Scholar 

  60. Cheng, X. & Ferrell, J. E. Jr. Apoptosis propagates through the cytoplasm as trigger waves. Science 361, 607–612 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Garcia-Perez, C. et al. Bid-induced mitochondrial membrane permeabilization waves propagated by local reactive oxygen species (ROS) signaling. Proc. Natl Acad. Sci. USA 109, 4497–4502 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Munoz-Pinedo, C. et al. Different mitochondrial intermembrane space proteins are released during apoptosis in a manner that is coordinately initiated but can vary in duration. Proc. Natl Acad. Sci. USA 103, 11573–11578 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Scorrano, L. et al. A distinct pathway remodels mitochondrial cristae and mobilizes cytochrome c during apoptosis. Dev. Cell 2, 55–67 (2002).

    CAS  PubMed  Google Scholar 

  64. Cogliati, S. et al. Mitochondrial cristae shape determines respiratory chain supercomplexes assembly and respiratory efficiency. Cell 155, 160–171 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Yamaguchi, R. et al. Opa1-mediated cristae opening is Bax/Bak and BH3 dependent, required for apoptosis, and independent of Bak oligomerization. Mol. Cell 31, 557–569 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Frank, S. et al. The role of dynamin-related protein 1, a mediator of mitochondrial fission, in apoptosis. Dev. Cell 1, 515–525 (2001).

    CAS  PubMed  Google Scholar 

  67. Frezza, C. et al. OPA1 controls apoptotic cristae remodeling independently from mitochondrial fusion. Cell 126, 177–189 (2006).

    CAS  PubMed  Google Scholar 

  68. van der Laan, M., Horvath, S. E. & Pfanner, N. Mitochondrial contact site and cristae organizing system. Curr. Opin. Cell Biol. 41, 33–42 (2016).

    PubMed  Google Scholar 

  69. Prudent, J. et al. MAPL SUMOylation of Drp1 stabilizes an ER/mitochondrial platform required for cell death. Mol. Cell 59, 941–955 (2015).

    CAS  PubMed  Google Scholar 

  70. Pernas, L. & Scorrano, L. Mito-morphosis: mitochondrial fusion, fission, and cristae remodeling as key mediators of cellular function. Annu. Rev. Physiol. 78, 505–531 (2016).

    CAS  PubMed  Google Scholar 

  71. Parone, P. A. et al. Inhibiting the mitochondrial fission machinery does not prevent Bax/Bak-dependent apoptosis. Mol. Cell Biol 26, 7397–7408 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Estaquier, J. & Arnoult, D. Inhibiting Drp1-mediated mitochondrial fission selectively prevents the release of cytochrome c during apoptosis. Cell Death Differ. 14, 1086–1094 (2007).

    CAS  PubMed  Google Scholar 

  73. Jiang, X., Jiang, H., Shen, Z. & Wang, X. Activation of mitochondrial protease OMA1 by Bax and Bak promotes cytochrome c release during apoptosis. Proc. Natl Acad. Sci. USA 111, 14782–14787 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Korwitz, A. et al. Loss of OMA1 delays neurodegeneration by preventing stress-induced OPA1 processing in mitochondria. J. Cell Biol. 212, 157–166 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Otera, H., Miyata, N., Kuge, O. & Mihara, K. Drp1-dependent mitochondrial fission via MiD49/51 is essential for apoptotic cristae remodeling. J. Cell Biol. 212, 531–544 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Sun, M. G. et al. Correlated three-dimensional light and electron microscopy reveals transformation of mitochondria during apoptosis. Nat. Cell Biol. 9, 1057–1065 (2007).

    CAS  PubMed  Google Scholar 

  77. Colell, A. et al. GAPDH and autophagy preserve survival after apoptotic cytochrome c release in the absence of caspase activation. Cell 129, 983–997 (2007).

    CAS  PubMed  Google Scholar 

  78. Tait, S. W. et al. Resistance to caspase-independent cell death requires persistence of intact mitochondria. Dev. Cell 18, 802–813 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Deshmukh, M. & Johnson, E. M. Jr. Evidence of a novel event during neuronal death: development of competence-to-die in response to cytoplasmic cytochrome c. Neuron 21, 695–705 (1998).

    CAS  PubMed  Google Scholar 

  80. Martinou, I. et al. The release of cytochrome c from mitochondria during apoptosis of NGF-deprived sympathetic neurons is a reversible event. J. Cell Biol. 144, 883–889 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Ichim, G. et al. Limited mitochondrial permeabilization causes DNA damage and genomic instability in the absence of cell death. Mol. Cell 57, 860–872 (2015). This study finds that MOMP can occur in a limited number of mitochondria within a cell, causing caspase activation without cell death.

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Gama, V. et al. The E3 ligase PARC mediates the degradation of cytosolic cytochrome c to promote survival in neurons and cancer cells. Sci. Signal 7, ra67 (2014).

    PubMed  PubMed Central  Google Scholar 

  83. Malladi, S., Challa-Malladi, M., Fearnhead, H. O. & Bratton, S. B. The Apaf-1*procaspase-9 apoptosome complex functions as a proteolytic-based molecular timer. EMBO J. 28, 1916–1925 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Kavanagh, E., Rodhe, J., Burguillos, M. A., Venero, J. L. & Joseph, B. Regulation of caspase-3 processing by cIAP2 controls the switch between pro-inflammatory activation and cell death in microglia. Cell Death Dis. 5, e1565 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Gonzalvez, F. et al. TRAF2 sets a threshold for extrinsic apoptosis by tagging caspase-8 with a ubiquitin shutoff timer. Mol. Cell 48, 888–899 (2012).

    CAS  PubMed  Google Scholar 

  86. Weber, G. F. & Menko, A. S. The canonical intrinsic mitochondrial death pathway has a non-apoptotic role in signaling lens cell differentiation. J. Biol. Chem. 280, 22135–22145 (2005).

    CAS  PubMed  Google Scholar 

  87. Ichim, G. & Tait, S. W. A fate worse than death: apoptosis as an oncogenic process. Nat. Rev. Cancer 16, 539–548 (2016).

    CAS  PubMed  Google Scholar 

  88. Liu, X. et al. Caspase-3 promotes genetic instability and carcinogenesis. Mol. Cell 58, 284–296 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  89. Lovric, M. M. & Hawkins, C. J. TRAIL treatment provokes mutations in surviving cells. Oncogene 29, 5048–5060 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Miles, M. A. & Hawkins, C. J. Executioner caspases and CAD are essential for mutagenesis induced by TRAIL or vincristine. Cell Death Dis. 8, e3062 (2017).

    PubMed  PubMed Central  Google Scholar 

  91. Cartwright, I. M., Liu, X., Zhou, M., Li, F. & Li, C. Y. Essential roles of caspase-3 in facilitating Myc-induced genetic instability and carcinogenesis. eLife 6, e26371 (2017).

    PubMed  PubMed Central  Google Scholar 

  92. Gong, Y. N., Crawford, J. C., Heckmann, B. L. & Green, D. R. To the edge of cell death and back. FEBS J. 286, 430–440 (2019).

    CAS  PubMed  Google Scholar 

  93. McArthur, K. & Kile, B. T. Apoptotic caspases: multiple or mistaken identities? Trends Cell Biol. 28, 475–493 (2018).

    CAS  PubMed  Google Scholar 

  94. Brokatzky, D. et al. A non-death function of the mitochondrial apoptosis apparatus in immunity. EMBO J. 38 (2019).

  95. Tang, H. L. et al. Cell survival, DNA damage, and oncogenic transformation after a transient and reversible apoptotic response. Mol. Biol. Cell 23, 2240–2252 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Sun, G. et al. A molecular signature for anastasis, recovery from the brink of apoptotic cell death. J. Cell Biol. 216, 3355–3368 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  97. Jiang, X. et al. A small molecule that protects the integrity of the electron transfer chain blocks the mitochondrial apoptotic pathway. Mol. Cell 63, 229–239 (2016).

    CAS  PubMed  Google Scholar 

  98. Martin, S. J., Henry, C. M. & Cullen, S. P. A perspective on mammalian caspases as positive and negative regulators of inflammation. Mol. Cell 46, 387–397 (2012).

    CAS  PubMed  Google Scholar 

  99. Green, D. R. Means to an end: apoptosis and other cell death mechanisms (Cold Spring Harbor Laboratory Press, 2010).

  100. Rongvaux, A. et al. Apoptotic caspases prevent the induction of type I interferons by mitochondrial DNA. Cell 159, 1563–1577 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  101. White, M. J. et al. Apoptotic caspases suppress mtDNA-induced STING-mediated type I IFN production. Cell 159, 1549–1562 (2014). Together with Rongvaux et al. (2014), this article demonstrates that under caspase-inhibited conditions MOMP activates cGAS–STING signalling dependent on mtDNA.

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Ablasser, A. & Chen, Z. J. cGAS in action: expanding roles in immunity and inflammation. Science 363, eaat8657 (2019).

    CAS  PubMed  Google Scholar 

  103. Riley, J. S. et al. Mitochondrial inner membrane permeabilisation enables mtDNA release during apoptosis. EMBO J. 37, e99238 (2018).

    PubMed  PubMed Central  Google Scholar 

  104. McArthur, K. et al. BAK/BAX macropores facilitate mitochondrial herniation and mtDNA efflux during apoptosis. Science 359, e99238 (2018).

    Google Scholar 

  105. Ader, N. R. et al. Molecular and topological reorganizations in mitochondrial architecture interplay during Bax-mediated steps of apoptosis. eLife 8, e40712 (2019). Together with Riley et al. (2018) and McArthur et al. (2018), this article describes the formation of BAX/BAK-dependent macropores on the mitochondrial outer membrane leading to inner mitochondrial membrane extrusion and mtDNA release.

    PubMed  PubMed Central  Google Scholar 

  106. Sliter, D. A. et al. Parkin and PINK1 mitigate STING-induced inflammation. Nature 561, 258–262 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  107. West, A. P. et al. Mitochondrial DNA stress primes the antiviral innate immune response. Nature 520, 553–557 (2015).

    PubMed  PubMed Central  Google Scholar 

  108. Aarreberg, L. D. et al. Interleukin-1β induces mtDNA release to activate innate immune signaling via cGAS-STING. Mol. Cell 74, 801–815 e806 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  109. Zhong, Z. et al. New mitochondrial DNA synthesis enables NLRP3 inflammasome activation. Nature 560, 198–203 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  110. Giampazolias, E. et al. Mitochondrial permeabilization engages NF-κB-dependent anti-tumour activity under caspase deficiency. Nat. Cell Biol. 19, 1116–1129 (2017). This study shows that under caspase-inhibited conditions MOMP elicits anti-tumour immunity, thereby supporting the rationale for inhibiting apoptotic caspase function in cancer.

    CAS  PubMed  PubMed Central  Google Scholar 

  111. Vince, J. E. et al. IAP antagonists target cIAP1 to induce TNFα-dependent apoptosis. Cell 131, 682–693 (2007).

    CAS  PubMed  Google Scholar 

  112. Varfolomeev, E. et al. IAP antagonists induce autoubiquitination of c-IAPs, NF-κB activation, and TNFα-dependent apoptosis. Cell 131, 669–681 (2007).

    CAS  PubMed  Google Scholar 

  113. Verhagen, A. M. et al. Identification of mammalian mitochondrial proteins that interact with IAPs via N-terminal IAP binding motifs. Cell Death Differ. 14, 348–357 (2007).

    CAS  PubMed  Google Scholar 

  114. Zhuang, M., Guan, S., Wang, H., Burlingame, A. L. & Wells, J. A. Substrates of IAP ubiquitin ligases identified with a designed orthogonal E3 ligase, the NEDDylator. Mol. Cell 49, 273–282 (2013).

    CAS  PubMed  Google Scholar 

  115. Chauhan, D. et al. BAX/BAK-induced apoptosis results in caspase-8-dependent IL-1β maturation in macrophages. Cell Rep. 25, 2354–2368 e2355 (2018).

    CAS  PubMed  Google Scholar 

  116. Vince, J. E. et al. The mitochondrial apoptotic effectors BAX/BAK activate caspase-3 and -7 to trigger NLRP3 inflammasome and caspase-8 driven IL-1β activation. Cell Rep. 25, 2339–2353 e2334 (2018).

    CAS  PubMed  Google Scholar 

  117. Tenev, T. et al. The ripoptosome, a signaling platform that assembles in response to genotoxic stress and loss of IAPs. Mol. Cell 43, 432–448 (2011).

    CAS  PubMed  Google Scholar 

  118. Feoktistova, M. et al. cIAPs block ripoptosome formation, a RIP1/caspase-8 containing intracellular cell death complex differentially regulated by cFLIP isoforms. Mol. Cell 43, 449–463 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  119. Chen, K. W. et al. Extrinsic and intrinsic apoptosis activate pannexin-1 to drive NLRP3 inflammasome assembly. EMBO J. 38, e101638 (2019).

    PubMed  PubMed Central  Google Scholar 

  120. Dhir, A. et al. Mitochondrial double-stranded RNA triggers antiviral signalling in humans. Nature 560, 238–242 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  121. Ning, X. et al. Apoptotic caspases suppress type I interferon production via the cleavage of cGAS, MAVS, and IRF3. Mol. Cell 74, 19–31 e17 (2019).

    CAS  PubMed  Google Scholar 

  122. Arandjelovic, S. & Ravichandran, K. S. Phagocytosis of apoptotic cells in homeostasis. Nat. Immunol. 16, 907–917 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  123. Leonard, J. R., Klocke, B. J., D’Sa, C., Flavell, R. A. & Roth, K. A. Strain-dependent neurodevelopmental abnormalities in caspase-3-deficient mice. J. Neuropathol. Exp. Neurol. 61, 673–677 (2002).

    PubMed  Google Scholar 

  124. Honarpour, N. et al. Adult Apaf-1-deficient mice exhibit male infertility. Dev. Biol. 218, 248–258 (2000).

    CAS  PubMed  Google Scholar 

  125. Liu, X. et al. PNPT1 release from mitochondria during apoptosis triggers decay of poly(A) RNAs. Cell 174, 187–201 e112 (2018).

    CAS  PubMed  Google Scholar 

  126. Lindqvist, L. M. et al. Autophagy induced during apoptosis degrades mitochondria and inhibits type I interferon secretion. Cell Death Differ. 25, 782–794 (2018).

    CAS  Google Scholar 

  127. Potts, M. B., Vaughn, A. E., McDonough, H., Patterson, C. & Deshmukh, M. Reduced Apaf-1 levels in cardiomyocytes engage strict regulation of apoptosis by endogenous XIAP. J. Cell Biol. 171, 925–930 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  128. Potts, P. R., Singh, S., Knezek, M., Thompson, C. B. & Deshmukh, M. Critical function of endogenous XIAP in regulating caspase activation during sympathetic neuronal apoptosis. J. Cell Biol. 163, 789–799 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  129. King, K. R. et al. IRF3 and type I interferons fuel a fatal response to myocardial infarction. Nat. Med. 23, 1481–1487 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  130. Thomas, R. L. et al. Loss of MCL-1 leads to impaired autophagy and rapid development of heart failure. Genes Dev. 27, 1365–1377 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  131. Messmer, M. N., Snyder, A. G. & Oberst, A. Comparing the effects of different cell death programs in tumor progression and immunotherapy. Cell Death Differ. 26, 115–129 (2019).

    PubMed  Google Scholar 

  132. Yatim, N., Cullen, S. & Albert, M. L. Dying cells actively regulate adaptive immune responses. Nat. Rev. Immunol. 17, 262–275 (2017).

    CAS  PubMed  Google Scholar 

  133. Kim, K. W., Moretti, L. & Lu, B. M867, a novel selective inhibitor of caspase-3 enhances cell death and extends tumor growth delay in irradiated lung cancer models. PLOS ONE 3, e2275 (2008).

    PubMed  PubMed Central  Google Scholar 

  134. Werthmoller, N., Frey, B., Wunderlich, R., Fietkau, R. & Gaipl, U. S. Modulation of radiochemoimmunotherapy-induced B16 melanoma cell death by the pan-caspase inhibitor zVAD-fmk induces anti-tumor immunity in a HMGB1-, nucleotide- and T-cell-dependent manner. Cell Death Dis. 6, e1761 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  135. Xu, M. et al. Identification of small-molecule inhibitors of Zika virus infection and induced neural cell death via a drug repurposing screen. Nat. Med. 22, 1101–1107 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  136. Weinlich, R., Oberst, A., Beere, H. M. & Green, D. R. Necroptosis in development, inflammation and disease. Nat. Rev. Mol. Cell Biol. 18, 127–136 (2017).

    CAS  PubMed  Google Scholar 

  137. Tait, S. W. et al. Widespread mitochondrial depletion via mitophagy does not compromise necroptosis. Cell Rep. 5, 878–885 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  138. Schenk, B. & Fulda, S. Reactive oxygen species regulate Smac mimetic/TNFα-induced necroptotic signaling and cell death. Oncogene 34, 5796–5806 (2015).

    CAS  PubMed  Google Scholar 

  139. Zhang, Y. et al. RIP1 autophosphorylation is promoted by mitochondrial ROS and is essential for RIP3 recruitment into necrosome. Nat. Commun. 8, 14329 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  140. Yang, Z. et al. RIP3 targets pyruvate dehydrogenase complex to increase aerobic respiration in TNF-induced necroptosis. Nat. Cell Biol. 20, 186–197 (2018).

    CAS  PubMed  Google Scholar 

  141. Broz, P. & Dixit, V. M. Inflammasomes: mechanism of assembly, regulation and signalling. Nat. Rev. Immunol. 16, 407–420 (2016).

    CAS  PubMed  Google Scholar 

  142. Kayagaki, N. et al. Caspase-11 cleaves gasdermin D for non-canonical inflammasome signalling. Nature 526, 666–671 (2015).

    CAS  PubMed  Google Scholar 

  143. Shi, J. et al. Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death. Nature 526, 660–665 (2015). Together with Kayagaki et al. (2015), this article demonstrates that caspase cleavage of gasdermin D causes plasma membrane permeabilization and pyroptosis.

    CAS  PubMed  Google Scholar 

  144. de Vasconcelos, N. M., Van Opdenbosch, N., Van Gorp, H., Parthoens, E. & Lamkanfi, M. Single-cell analysis of pyroptosis dynamics reveals conserved GSDMD-mediated subcellular events that precede plasma membrane rupture. Cell Death Differ. 26, 146–161 (2019).

    PubMed  Google Scholar 

  145. Tsuchiya, K. et al. Caspase-1 initiates apoptosis in the absence of gasdermin D. Nat. Commun. 10, 2091 (2019).

    PubMed  PubMed Central  Google Scholar 

  146. Stockwell, B. R. et al. Ferroptosis: a regulated cell death nexus linking metabolism, redox biology, and disease. Cell 171, 273–285 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  147. Dixon, S. J. et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell 149, 1060–1072 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  148. Yang, W. S. et al. Regulation of ferroptotic cancer cell death by GPX4. Cell 156, 317–331 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  149. Gaschler, M. M. et al. Determination of the subcellular localization and mechanism of action of ferrostatins in suppressing ferroptosis. ACS Chem. Biol. 13, 1013–1020 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  150. Wang, Y. Q. et al. The protective role of mitochondrial ferritin on erastin-induced ferroptosis. Front. Aging Neurosci. 8, 308 (2016).

    PubMed  PubMed Central  Google Scholar 

  151. Fang, X. X. et al. Ferroptosis as a target for protection against cardiomyopathy. Proc. Natl Acad. Sci. USA 116, 2672–2680 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  152. Gao, M. H. et al. Role of mitochondria in ferroptosis. Mol. Cell 73, 354–363.e3 (2019).

    CAS  PubMed  Google Scholar 

  153. Ding, A. X. et al. CasExpress reveals widespread and diverse patterns of cell survival of caspase-3 activation during development in vivo. eLife 5, e10936 (2016).

    PubMed  PubMed Central  Google Scholar 

  154. Lagares, D. et al. Targeted apoptosis of myofibroblasts with the BH3 mimetic ABT-263 reverses established fibrosis. Sci. Transl. Med. 9, eaal3765 (2017).

    PubMed  PubMed Central  Google Scholar 

  155. Chang, J. et al. Clearance of senescent cells by ABT263 rejuvenates aged hematopoietic stem cells in mice. Nat. Med. 22, 78–83 (2016).

    CAS  PubMed  Google Scholar 

  156. Garner, T. P. et al. Small-molecule allosteric inhibitors of BAX. Nat. Chem. Biol. 15, 322–330 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  157. Niu, X. et al. A small-molecule inhibitor of bax and bak oligomerization prevents genotoxic cell death and promotes neuroprotection. Cell Chem. Biol. 24, 493–506 e495 (2017). Together with Garner et al. (2019), this work describes the development of small-molecule BAX/BAK inhibitors that may serve as prototypes to develop therapeutic inhibitors of mitochondrial apoptosis.

    CAS  PubMed  PubMed Central  Google Scholar 

  158. Singh, R., Letai, A. & Sarosiek, K. Regulation of apoptosis in health and disease: the balancing act of BCL-2 family proteins. Nat. Rev. Mol. Cell Biol. 20, 175–193 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  159. Llambi, F. et al. A unified model of mammalian BCL-2 protein family interactions at the mitochondria. Mol. Cell 44, 517–531 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  160. Pecot, J. et al. Tight sequestration of BH3 proteins by BCL-xL at subcellular membranes contributes to apoptotic resistance. Cell Rep. 17, 3347–3358 (2016).

    CAS  PubMed  Google Scholar 

  161. Liu, Q. et al. Bim escapes displacement by BH3-mimetic anti-cancer drugs by double-bolt locking both Bcl-XL and Bcl-2. eLife 8, e37689 (2019).

    PubMed  PubMed Central  Google Scholar 

  162. O’Neill, K. L., Huang, K., Zhang, J., Chen, Y. & Luo, X. Inactivation of prosurvival Bcl-2 proteins activates Bax/Bak through the outer mitochondrial membrane. Genes Dev. 30, 973–988 (2016). This study shows that in the absence of all known BH3-only proteins, inhibition of anti-apoptotic BCL-2 proteins is sufficient to activate BAX and BAK.

    PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank S. Dixon and T. Mouldoveanu for discussion and critical input. Research in the authors’ laboratory is supported by funding from Cancer Research UK (C40872/A20145) and Prostate Cancer UK (RIA17-ST2-002).

Author information

Authors and Affiliations

Authors

Contributions

The authors contributed equally to all aspects of the article.

Corresponding author

Correspondence to Stephen W. G. Tait.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review informationNature Reviews Molecular Cell Biology thanks P. Juin and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Glossary

BH3-mimetics

Drugs modelled after the pro-apoptotic BH3 domain of BH3-only proteins that are used in cancer therapy.

Death-inducing signalling complex

(DISC). A complex consisting of death receptor, Fas-associated death domain (FADD) and caspase 8 that can mediate apoptosis.

SMAC

(Also known as DIABLO). A mitochondrial intermembrane space protein that upon mitochondrial outer membrane permeabilization binds to and inhibits XIAP.

OMI

(Also known as HtrA2). A serine protease located within the mitochondrial intermembrane space that binds to and inhibits XIAP following mitochondrial outer membrane permeabilization.

XIAP

A protein that binds to and inhibits caspases 3, 7 and 9.

Endoplasmic reticulum-associated degradation

Pathway that serves to degrade misfolded endoplasmic reticulum (ER) proteins by the proteasome, mitigating ER stress.

Neoantigens

Newly generated antigens that, in cancer, usually arise from mutated genes.

Type I interferon

Class of cytokines mediating inflammation.

SMAC-mimetic compounds

Chemicals that were designed to phenocopy the inhibitor of apoptosis protein-binding and inhibitory properties of SMAC.

NLRP3 inflammasome

A protein complex containing NOD-, LRR- and pyrin domain-containing 3 (NLRP3) and caspase 1 that processes and activates inflammatory cytokines such as IL-1β and IL-18.

RNA degradasome

A multiprotein complex present in bacteria and mitochondria that degrades RNA.

‘Find-me’ and ‘eat-me’ signals

Molecular signals used by dying cells to attract phagocytes; examples of find-me signals include ATP and lysophosphatidylcholine, and the best-characterized eat-me signal is phosphatidylserine.

Ischaemic injury

Hypoxia-mediated injury due to diminished blood flow.

Toll receptor

A class of protein receptors that serve a key role in innate immunity by sensing conserved molecules derived from microorganisms.

Necrosome

A protein complex containing receptor interacting protein kinase 1 (RIPK1) and RIPK3 that promotes necroptotic cell death.

Fenton reaction

The reaction of peroxides with iron to yield free radicals.

Glutathione

A key cellular antioxidant that scavenges reactive oxygen species through reduction.

Ferritin

An iron-binding protein that plays important roles in the storage and transport of iron throughout the body.

Haeme

An iron-containing coordination complex present in haemoproteins such as haemoglobin, catalases and cytochrome c.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Bock, F.J., Tait, S.W.G. Mitochondria as multifaceted regulators of cell death. Nat Rev Mol Cell Biol 21, 85–100 (2020). https://doi.org/10.1038/s41580-019-0173-8

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41580-019-0173-8

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing